Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bradley P. Sullivan is active.

Publication


Featured researches published by Bradley P. Sullivan.


Molecular Microbiology | 2012

FtsA mutants impaired for self‐interaction bypass ZipA suggesting a model in which FtsA's self‐interaction competes with its ability to recruit downstream division proteins

Sebastien Pichoff; Bang Shen; Bradley P. Sullivan; Joe Lutkenhaus

Z‐ring assembly requires polymers of the tubulin homologue FtsZ to be tethered to the membrane. Although either ZipA or FtsA is sufficient to do this, both of these are required for recruitment of downstream proteins to form a functional cytokinetic ring. Gain of function mutations in ftsA, such as ftsA* (ftsA‐R286W), bypass the requirement for ZipA suggesting that this atypical, well‐conserved, actin homologue has a more critical role in Z‐ring function. FtsA forms multimers both in vitro and in vivo, but little is known about the role of FtsA polymerization. In this study we identify FtsA mutants impaired for self‐interaction. Such mutants are able to support Z‐ring assembly and are also able to bypass the requirement for ZipA. These mutants, including FtsA*, have reduced ability to self‐interact but interact normally with FtsZ and are less toxic if overexpressed. These results do not support a model in which FtsA monomers antagonize FtsZ polymers. Instead, we propose a new model in which FtsA self‐interaction competes with its ability to recruit downstream proteins. In this model FtsA self‐interaction at the Z ring is antagonized by ZipA, allowing unpolymerized FtsA to recruit downstream proteins such as FtsN.


American Journal of Pathology | 2010

The Coagulation System Contributes to αVβ6 Integrin Expression and Liver Fibrosis Induced by Cholestasis

Bradley P. Sullivan; Paul H. Weinreb; Shelia M. Violette; James P. Luyendyk

Chronic injury to intrahepatic bile duct epithelial cells (BDECs) elicits expression of various mediators, including the αVβ6 integrin, promoting liver fibrosis. We tested the hypothesis that tissue factor (TF)-dependent thrombin generation and protease activated receptor-1 (PAR-1) activation contribute to liver fibrosis induced by cholestasis via induction of αVβ6 expression. To test this hypothesis, mice deficient in either TF or PAR-1 were fed a diet containing 0.025% α-naphthylisothiocyanate (ANIT), a BDEC-selective toxicant. In genetically modified mice with a 50% reduction in liver TF activity fed an ANIT diet, coagulation cascade activation and liver fibrosis were reduced. Similarly, liver fibrosis was significantly reduced in PAR-1(-/-) mice fed an ANIT diet. Hepatic integrin β6 mRNA induction, expression of αVβ6 protein by intrahepatic BDECs, and SMAD2 phosphorylation were reduced by TF deficiency and PAR-1 deficiency in mice fed the ANIT diet. Treatment with either an anti-αVβ6 blocking antibody or soluble transforming growth factor-β receptor type II reduced liver fibrosis in mice fed the ANIT diet. PAR-1 activation enhanced transforming growth factor-β1-induced integrin β6 mRNA expression in both transformed human BDECs and primary rat BDECs. Interestingly, TF and PAR-1 mRNA levels were increased in livers from patients with cholestatic liver disease. These results indicate that a TF-PAR-1 pathway contributes to liver fibrosis induced by chronic cholestasis by increasing expression of the αVβ6 integrin, an important regulator of transforming growth factor-β1 activation.


American Journal of Pathology | 2010

Tissue factor-deficiency and protease activated receptor-1-deficiency reduce inflammation elicited by diet-induced steatohepatitis in mice.

James P. Luyendyk; Bradley P. Sullivan; Grace L. Guo; Ruipeng Wang

Altered hepatic lipid homeostasis, hepatocellular injury, and inflammation are features of nonalcoholic steatohepatitis, which contributes significantly to liver-related morbidity and mortality in the Western population. A collection of inflammatory mediators have been implicated in the pathogenesis of steatohepatitis in mouse models. However, the pathways essential for coordination and amplification of hepatic inflammation and injury caused by steatosis are not completely understood. We tested the hypothesis that tissue factor (TF)-dependent thrombin generation and the thrombin receptor protease activated receptor-1 (PAR-1) contribute to liver inflammation induced by steatosis in mice. Wild-type C57Bl/6J mice fed a diet deficient in methionine and choline for 2 weeks manifested steatohepatitis characterized by increased serum alanine aminotransferase activity, macrovesicular hepatic steatosis, hepatic inflammatory gene expression, and lobular inflammation. Steatohepatitis progression was associated with thrombin generation and hepatic fibrin deposition. Coagulation cascade activation was significantly reduced in low TF mice, which express 1% of normal TF levels. Hepatic triglyceride accumulation was not affected in low TF mice or PAR-1-deficient mice. In contrast, biomarkers of hepatocellular injury, inflammatory gene induction, and hepatic accumulation of macrophages and neutrophils were greatly reduced by TF-deficiency and PAR-1-deficiency. The results suggest that TF-dependent thrombin generation and activation of PAR-1 amplify hepatic inflammation and injury during the pathogenesis of steatohepatitis.


Blood | 2013

Hepatocyte tissue factor activates the coagulation cascade in mice

Bradley P. Sullivan; Anna K. Kopec; Nikita Joshi; Holly Cline; Juliette A. Brown; Stephanie C. Bishop; Karen M. Kassel; Cheryl E. Rockwell; Nigel Mackman; James P. Luyendyk

In this study, we characterized tissue factor (TF) expression in mouse hepatocytes (HPCs) and evaluated its role in mouse models of HPC transplantation and acetaminophen (APAP) overdose. TF expression was significantly reduced in isolated HPCs and liver homogenates from TF(flox/flox)/albumin-Cre mice (HPC(ΔTF) mice) compared with TF(flox/flox) mice (control mice). Isolated mouse HPCs expressed low levels of TF that clotted factor VII-deficient human plasma. In addition, HPC TF initiated factor Xa generation without exogenous factor VIIa, and TF activity was increased dramatically after cell lysis. Treatment of HPCs with an inhibitory TF antibody or a cell-impermeable lysine-conjugating reagent prior to lysis substantially reduced TF activity, suggesting that TF was mainly present on the cell surface. Thrombin generation was dramatically reduced in APAP-treated HPC(ΔTF) mice compared with APAP-treated control mice. In addition, thrombin generation was dependent on donor HPC TF expression in a model of HPC transplantation. These results suggest that mouse HPCs constitutively express cell surface TF that mediates activation of coagulation during hepatocellular injury.


Toxicological Sciences | 2010

Protective and Damaging Effects of Platelets in Acute Cholestatic Liver Injury Revealed by Depletion and Inhibition Strategies

Bradley P. Sullivan; Ruipeng Wang; Ossama Tawfik; James P. Luyendyk

Alpha-naphthylisothiocyanate (ANIT) causes cholestatic hepatitis characterized by intrahepatic bile duct epithelial cell injury and periportal hepatocellular necrosis. The progression of ANIT-induced hepatocyte injury is reported to involve extrahepatic cells including platelets. We showed recently that the procoagulant protein tissue factor (TF) is essential for ANIT-induced coagulation and contributes to ANIT-induced liver necrosis. Platelets have been shown to express TF and can contribute to coagulation cascade activation. To this end, we tested the hypothesis that platelet-dependent coagulation contributes to ANIT-induced liver injury. In ANIT (60 mg/kg)-treated mice, activation of the coagulation cascade occurred prior to a decrease of platelets in the blood. Immunostaining for glycoprotein IIb (CD41) revealed platelet accumulation along the borders of necrotic foci in livers of ANIT-treated mice. Antibody-mediated platelet depletion did not affect coagulation but markedly affected liver histopathology in ANIT-treated mice. Platelet depletion induced marked pooling of blood within necrotic lesions consistent with parenchymal-type peliosis as early as 24 h after ANIT treatment. In contrast, treatment with the P2Y(12) inhibitor clopidogrel significantly reduced ANIT-induced hepatocyte necrosis and serum alanine aminotransferase activity but did not exaggerate bleeding into necrotic foci. Clopidogrel also reduced hepatic neutrophil accumulation but did not affect induction of Intercellular adhesion molecule-1 or chemokine CxC motif ligand-1 messenger RNA expression in liver. The data indicate that ANIT-induced coagulation is platelet independent and that platelets contribute to ANIT-induced hepatocyte necrosis by promoting neutrophil accumulation. In contrast, severe thrombocytopenia induces parenchymal-type peliosis in the livers of ANIT-treated mice, a rare hepatic lesion associated with pooling of blood in the liver.


Advanced Drug Delivery Reviews | 2016

Combining antigen and immunomodulators: Emerging trends in antigen-specific immunotherapy for autoimmunity.

Laura Northrup; Matthew A. Christopher; Bradley P. Sullivan; Cory Berkland

A majority of current therapies for autoimmune diseases are general immunosuppressants, which can compromise patient response to opportunistic infection and lead to adverse events. Using antigen-specific immunotherapy (ASIT) to selectively disarm autoimmune diseases, without suppressing the global immune response, would be a transformative therapy for patients. ASIT has been used historically in allergy hyposensitization therapy to induce tolerance to an allergen. Similar strategies to induce immune tolerance toward autoantigens responsible for autoimmune disease have been attempted but have yielded limited clinical success. Recent studies of ASIT for autoimmunity have explored combination therapy, combining the disease-causing autoantigen with an immunomodulatory compound. ASIT combination therapy may direct the immune response in an antigen-specific manner, potentially reversing the root cause of autoimmunity while limiting side effects. This review analyzes recent advances in ASIT applied to autoimmune diseases, emphasizing current combination therapies and future strategies.


American Journal of Pathology | 2012

Therapeutic Administration of the Direct Thrombin Inhibitor Argatroban Reduces Hepatic Inflammation in Mice with Established Fatty Liver Disease

Karen M. Kassel; Bradley P. Sullivan; Wei Cui; Bryan L. Copple; James P. Luyendyk

Thrombin generation is increased in patients with nonalcoholic fatty liver disease (NAFLD) and in mouse models of diet-induced obesity. Deficiency in the thrombin receptor protease activated receptor-1 reduces hepatic inflammation and steatosis in mice fed a Western diet. However, it is currently unclear whether thrombin inhibitors can modify the pathogenesis of established NAFLD. We tested the hypothesis that thrombin inhibition could reverse hepatic steatosis and inflammation in mice with established diet-induced NAFLD. Low-density lipoprotein receptor-deficient LDLr(-/-) mice were fed a control diet or a Western diet for 19 weeks. Mice were given the direct thrombin inhibitor argatroban ∼15 mg/kg/day or its vehicle via a miniosmotic pump for the final 4 weeks of the study. Argatroban administration significantly reduced hepatic proinflammatory cytokine expression and reduced macrophage and neutrophil accumulation in livers of mice fed a Western diet. Argatroban did not significantly impact hepatic steatosis, as indicated by histopathology, Oil Red O staining, and hepatic triglyceride levels. Argatroban reduced serum triglyceride and cholesterol levels in mice fed a Western diet. Argatroban reduced both α-smooth muscle actin expression and Type 1 collagen mRNA levels in livers of mice fed a Western diet, indicating reduced activation of hepatic stellate cells. This study indicates that therapeutic intervention with a thrombin inhibitor attenuates hepatic inflammation and several profibrogenic changes in mice fed a Western diet.


Journal of Pharmaceutical Sciences | 2015

Multivalent Nanomaterials: Learning from Vaccines and Progressing to Antigen‐Specific Immunotherapies

Brittany L. Hartwell; Lorena Antunez; Bradley P. Sullivan; Sharadvi Thati; Joshua Sestak; Cory Berkland

Continued development of multivalent nanomaterials has provided opportunities for the advancement of antigen-specific immunotherapies. New insights emerge when considering the backdrop of vaccine design, which has long employed multivalent presentation of antigen to more strongly engage and enhance an immunogenic response. Additionally, vaccines traditionally codeliver antigen with adjuvant to amplify a robust antigen-specific response. Multivalent nanomaterials have since evolved for applications where immune tolerance is desired, such as autoimmune diseases or allergies. In particular, soluble, linear polymers may be tailored to direct antigen-specific immunogenicity or tolerance by modulating polymer length, ligand valency (number), and ligand density, in addition to incorporating secondary signals. Codelivery of a secondary signal may direct, amplify, or suppress the response to a given antigen. Although the ability of multivalent nanomaterials to enact an immune response through molecular mechanisms has been established, a transport mechanism for biodistribution must also be considered. Both mechanisms are influenced by ligand display and other physical properties of the nanomaterial. This review highlights multivalent ligand display on linear polymers, the complex interplay of physical parameters in multivalent design, and the ability to direct the immune response by molecular and transport mechanisms.


American Journal of Pathology | 2012

Fibrin(ogen)-Independent Role of Plasminogen Activators in Acetaminophen-Induced Liver Injury

Bradley P. Sullivan; Karen M. Kassel; Alice Jone; Matthew J. Flick; James P. Luyendyk

Hepatic fibrin(ogen) has been noted to occur after acetaminophen (APAP)-induced liver injury in mice. Deficiency in plasminogen activator inhibitor-1 (PAI-1), an endogenous inhibitor of fibrinolysis, increases APAP-induced liver injury in mice. However, the roles of fibrinogen and fibrinolysis in APAP-induced liver injury are not known. We tested the hypothesis that hepatic fibrin(ogen) deposition reduces severity of APAP-induced liver injury. APAP-induced (300 mg/kg) liver injury in mice was accompanied by thrombin generation, consumption of plasma fibrinogen, and deposition of hepatic fibrin. Neither fibrinogen depletion with ancrod nor complete fibrinogen deficiency [via knockout of the fibrinogen alpha chain gene (Fbg(-/-))] affected APAP-induced liver injury. PAI-1 deficiency (PAI-1(-/-)) increased APAP-induced liver injury and hepatic fibrin deposition 6 hours after APAP administration, which was followed by marked hemorrhage at 24 hours. As in PAI-1(-/-) mice, administration of recombinant tissue plasminogen activator (tenecteplase, 5 mg/kg) worsened APAP-induced liver injury and hemorrhage in wild-type mice. In contrast, APAP-induced liver injury was reduced in both plasminogen-deficient mice and in wild-type mice treated with tranexamic acid, an inhibitor of plasminogen activation. Activation of matrix metalloproteinase 9 (MMP-9) paralleled injury, but MMP-9 deficiency did not affect APAP-induced liver injury. The results indicate that fibrin(ogen) does not contribute to development of APAP-induced liver injury and suggest rather that plasminogen activation contributes to APAP-induced liver injury.


American Journal of Pathology | 2011

Protease-Activated Receptor 1 and Hematopoietic Cell Tissue Factor Are Required for Hepatic Steatosis in Mice Fed a Western Diet

Karen M. Kassel; A. Phillip Owens; Cheryl E. Rockwell; Bradley P. Sullivan; Ruipeng Wang; Ossama Tawfik; Guodong Li; Grace L. Guo; Nigel Mackman; James P. Luyendyk

Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of obesity and metabolic syndrome and contributes to increased risk of cardiovascular disease and liver-related morbidity and mortality. Indeed, obese patients with metabolic syndrome generate greater amounts of thrombin, an indication of coagulation cascade activation. However, the role of the coagulation cascade in Western diet-induced NAFLD has not been investigated. Using an established mouse model of Western diet-induced NAFLD, we tested whether the thrombin receptor protease-activated receptor 1 (PAR-1) and hematopoietic cell-derived tissue factor (TF) contribute to hepatic steatosis. In association with hepatic steatosis, plasma thrombin-antithrombin levels and hepatic fibrin deposition increased significantly in C57Bl/6J mice fed a Western diet for 3 months. PAR-1 deficiency reduced hepatic inflammation, particularly monocyte chemoattractant protein-1 expression and macrophage accumulation. In addition, PAR-1 deficiency was associated with reduced steatosis in mice fed a Western diet, including reduced liver triglyceride accumulation and CD36 expression. Similar to PAR-1 deficiency, hematopoietic cell TF deficiency was associated with reduced inflammation and reduced steatosis in livers of low-density lipoprotein receptor-deficient mice fed a Western diet. Moreover, hematopoietic cell TF deficiency reduced hepatic fibrin deposition. These studies indicate that PAR-1 and hematopoietic cell TF are required for liver inflammation and steatosis in mice fed a Western diet.

Collaboration


Dive into the Bradley P. Sullivan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karen M. Kassel

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna K. Kopec

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nigel Mackman

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge